Download The Essential Role of Fibroblasts in Esophageal Squamous Cell

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts
no text concepts found
Transcript
GASTROENTEROLOGY 2008;134:1981–1993
The Essential Role of Fibroblasts in Esophageal Squamous Cell
Carcinoma–Induced Angiogenesis
KAZUHIRO NOMA,* KEIRAN S. M. SMALLEY,* MERCEDES LIONI,* YOSHIO NAOMOTO,‡ NORIAKI TANAKA,‡
WAFIK EL–DEIRY,§ ALASTAIR J. KING,储 HIROSHI NAKAGAWA,¶ and MEENHARD HERLYN*
Background & Aims: Esophageal squamous cell carcinoma (ESCC) is known to be a highly angiogenic
tumor. Here, we investigated the role of the stromal
fibroblasts in the ESCC-induced angiogenic response
using a novel 3-dimensional model. Methods: A novel
assay was developed where cocultures of ESCC and
esophageal fibroblasts induced human microvascular
endothelial cell (HMVEC) vascular network formation
in a 3-dimensional collagen gel. Biochemical studies
showed that the ESCC-induced activation of the fibroblasts was required to induce vascular network formation via a transforming growth factor (TGF)-␤ and vascular endothelial growth factor (VEGF)-dependent
pathway. Results: Conditioned media from a panel
of 4 ESCC lines transdifferentiated normal esophageal fibroblasts into myofibroblasts via TGF-␤ signaling. The presence of fibroblasts was essential for efficient HMVEC network formation, and the addition
of ESCC cells to these cultures greatly enhanced the
angiogenic process. The role of TGF-␤ in this process
was shown by the complete inhibition of network formation following TGF-␤ inhibitor treatment. Finally,
we showed that ESCC-derived TGF-␤ regulates angiogenesis through the release of VEGF from the fibroblasts and that the VEGF release was blocked following TGF-␤ inhibition. Conclusions: This study
shows the essential role of fibroblasts in the ESCC
angiogenic-induced response and suggests that the
pharmacologic targeting of the TGF-␤ signaling axis
could be of therapeutic benefit in this deadly disease.
T
he tumor “organ” consists of a dynamic mixture of
tumor cells, fibroblasts, endothelial cells, and immune cells that all work together to drive tumor progression.1 Activated fibroblasts, also known as carcinomaassociated fibroblasts (CAFs),2 have been identified at the
leading edges of many solid tumors, including breast,
colon, and melanoma.3–5 The presence of CAFs within
the tumor microenvironment is preceded by the chemoattraction and migration of precursor cells, which can
either arise from the surrounding host fibroblasts or
from circulating mesenchymal precursors/stem cells.6,7
Once recruited, paracrine tumor-derived growth factors activate the CAFs, which undergo a myofibroblastic transdifferentiation, defined by an elongated spindle shape, and the
expression of contractile ␣–smooth muscle actin (␣-SMA)
and vimentin.8 CAFs are hypothesized to drive tumor
progression through the deposition of extracellular matrix proteins, the secretion of growth factors, and the
stimulation of invasion.9
One area that has been little explored is the potential
role of CAFs in tumor angiogenesis. Much of the growth
of solid tumors is dependent on the ready supply of
nutrients and oxygen from a local blood supply. As tumors grow beyond a few millimeters in size, they readily
outstrip the local supply of nutrients available through
simple diffusion and stimulate the formation of their
own tumor vasculature. Although it has been shown that
stromal fibroblasts are an important source of the proangiogenic factor vascular endothelial cell growth factor
(VEGF),10 it has been difficult to study the interaction of
carcinoma cells, fibroblasts, and endothelial cells in a
physiologically relevant model. In the present study, we
used a novel 3-dimensional (3D) in vitro model in which
the interaction of esophageal squamous cell carcinoma
(ESCC) cells with fibroblasts drives vascular network formation in a 3D collagen gel. We show that ESCC cells
require the presence of stromal fibroblasts to stimulate
vascular network formation, thereby suggesting that fibroblasts are the critical mediators of angiogenesis in this
system. Mechanistic studies reveal that paracrine transforming growth factor (TGF)-␤ from the ESCC cells leads
to activation of the fibroblasts and that pharmacologic
Abbreviations used in this paper: CAF, carcinoma-associated fibroblast; DAPI, 4=,6-diamidino-2-phenylindole; DMEM, Dulbecco’s modified Eagle medium; ELISA, enzyme-linked immunosorbent assay;
ESCC, esophageal squamous cell carcinoma; FBS, fetal bovine serum;
GFP, green fluorescent protein; HMVEC, human microvascular endothelial cell; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium
bromide; ␣-SMA, ␣–smooth muscle actin; TGF, transforming growth
factor; 3D, 3-dimensional; VEGF, vascular endothelial growth factor.
© 2008 by the AGA Institute
0016-5085/08/$34.00
doi:10.1053/j.gastro.2008.02.061
BASIC–
ALIMENTARY TRACT
*The Wistar Institute, Philadelphia, Pennsylvania; ‡Departments of Gastroenterological Surgery, Transplant and Surgical Oncology, Graduate School of Medicine,
Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan; §Hematology-Oncology Division and ¶Gastroenterology Division, Department of
Medicine, Department of Genetics, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania; and 储GlaxoSmithKline,
Collegeville, Pennsylvania
1982
NOMA ET AL
inhibitors of TGF-␤ signaling can reverse both fibroblast
activation and vascular network formation.
Materials and Methods
Cell Lines
Esophageal cancer cells, TE cell lines (TE1, TE8,
TE10, TE11, TE12), were cultured as previously described.11 Human esophageal keratinocytes EPC2 have
been described previously.11,12 Human microvascular endothelial cells (HMVECs) are available commercially
through Cascade Biologics, Inc (Portland, OR).13 Primary
human esophageal fibroblasts designated as FEF3 were
isolated from human fetal esophagus as described previously.11 FEF3 cells were stably transduced using the ViraPower Lentiviral Expression System (Invitrogen, Carlsbad, CA) containing the gene for green fluorescent
protein (GFP). GFP lentivirus was raised in our laboratory, and the FEF3 cells were transduced in the presence
of 6 ␮g/mL polybrene. Forty-eight hours after transduction, cells were selected in the presence of 10 ␮g/mL
blasticidin for 14 days.
Antibodies and Reagents
BASIC–
ALIMENTARY TRACT
The following antibodies were used in this study:
anti-human CD31 (Dako, Carpinteria, CA), anti–␣-SMA
(Sigma-Aldrich, St Louis, MO), anti–von Willebrand factor (Neomarkers, Fremont, CA), anti–fibroblast activation protein (EMD Biosciences, San Diego, CA), antiTGF␤RII (Santa Cruz Biotechnology, Santa Cruz, CA),
and phalloidin/Texas Red (Molecular Probes, Eugene,
OR). The anti-smad2, anti–phospho-smad2 (Ser465/
467), anti–phospho-smad3 (Ser423/425), and Smad1
(Ser463/465) antibodies were purchased from Cell Signaling Technology (Beverly, MA). Recombinant human
TGF-␤1 was purchased by R&D Systems, Inc (Minneapolis, MN). SB-505124, GW788388, and GW654652 were
provided by GlaxoSmithKline (Collegeville, PA). Bevacizumab (Avastin; Genentech, South San Francisco, CA)
was obtained from the pharmacy of the Hospital of the
University of Pennsylvania.
In Vitro 3D Network Formation Assay and
Fluorescence Imaging
Reconstruction of a vessel-like structure in 3D
collagen gels and subsequent fluorescent staining of networks/cords in whole-mount gels were performed as previously described.13 Briefly, HMVECs were cultured as
monolayers on bovine type I collagen-coated 24-well
plates at 1.5 ⫻ 105 cells/well for 24 hours and overlaid
with acellular collagen mixed in 10⫻ Medium 199 (Invitrogen) with heparin (100 U/mL), vitamin C (50 ␮g/
mL), and fetal bovine serum (FBS; 1%). After polymerization of the collagen gels, the cells were further overlaid
with a second collagen layer containing each 1.0 –2.5 ⫻
105 cells/mL FEF3, 0.5 ⫻ 105 cells/mL TE cells, or both
cells. Wells were then filled with EBM-2 medium contain-
GASTROENTEROLOGY Vol. 134, No. 7
ing EGM-2MV. The reconstructs were incubated at 37°C
for 7 days. To prepare for staining, medium was removed
and the collagen gels were fixed in Prefer (Anatech Ltd,
Battle Creek, MI) for 4 hours at room temperature. Gels
were processed as whole mounts. After blocking with 1%
bovine serum albumin, gels were stained with monoclonal anti-CD31 antibody followed by Texas Red– conjugated secondary antibody. Gels were treated with
VectaShield with 4=,6-diamidino-2-phenylindole (DAPI)
(Vector Laboratories, Burlingame, CA), and the stained
endothelial cell networks were photographed under a
Nikon E600 fluorescent microscope (Nikon, Melville,
NY).
The capillary-like networks were scored by counting
the number of CD31-stained branches. One branch was
counted to be 3 cells thick or less (to discount disorganized masses of HMVECs) and at least 3 whole cells long.
At least 5 randomly chosen low-power fields were
counted per sample (10⫻ magnification). Each figure
shows one representative experiment. Data show the
mean of at least 3 independent experiments.
Immunofluorescence Microscope
For cocultures, FEF3 cells and TE cells mixed in a
1:1 ratio (2.5 ⫻ 104 cells per each) were seeded onto glass
coverslips in 6-well plates and cultured in Dulbecco’s
modified Eagle medium (DMEM) containing 10% FBS
for 48 hours. Cells were then fixed and stained as described previously.11
Western Blotting Analysis
Subconfluent cells were lysed and separated on a
4%–12% sodium dodecyl sulfate/polyacrylamide gel before being blotted as described previously.11
Treatment of FEF3 Cells With Conditioned
Media
For preparation of conditioned medium, TE cells
were cultured with DMEM containing 10% FBS overnight. Supernatants were removed, and cells were washed
with DMEM. Cells were cultured for 48 hours with fresh
medium DMEM containing 2% FBS. FEF3 cells were
cultured overnight with DMEM containing 10% FBS.
Supernatants were replaced and cultured with fresh culture medium DMEM containing 2% FBS with or without
TGF-␤1 or cultured with conditioned medium for each
time.
TGF-␤1/2 Enzyme-Linked Immunosorbent
Assay
Cells were cultured overnight with DMEM containing 10% FBS. Supernatants of these cells were removed, and cells were washed with DMEM basal medium. Cells were cultured for 48 hours with fresh culture
medium DMEM containing 2% FBS. These supernatants
were measured as samples using each enzyme-linked im-
FIBROBLASTS DRIVE ESCC–INDUCED ANGIOGENESIS
1983
BASIC–
ALIMENTARY TRACT
June 2008
Figure 1. The interaction of ESCC cells and fibroblasts drives efficient vascular network formation in 3D culture. Immunofluorescence staining shows GFPtagged FEF3 esophageal fibroblasts (green), HMVECs (CD31; red), and total nuclei (DAPI; blue). (Inset) CD31 staining. (A) Incubation of the HMVECs with each of
3 ESCC lines (TE1, TE8, TE11) was not associated with any vascular network formation. (B) Coculture of the HMVECs with human esophageal fibroblasts (FEF3)
led to increased vascular network formation. (C) The addition of ESCC cells to the fibroblast/endothelial cell coculture markedly increased the organization of the
vascular networks. Increasing the culture time to 14 days dramatically enhanced the organization of the vascular network. (D) ESCC number was also found to
increase the degree of vascular network formation. (E) The extent of network formation under each of the culture conditions was scored, with the addition of
increasing numbers of ESCC cells found to significantly increase the level of vascular network formation. Scale bar ⫽ 200 ␮m.
1984
NOMA ET AL
GASTROENTEROLOGY Vol. 134, No. 7
BASIC–
ALIMENTARY TRACT
munosorbent assay (ELISA). ELISA kits for TGF-␤1,
TGF-␤2, and VEGF were purchased from R&D Systems.
These assays were performed according to the manufacturer’s instructions. For TGF-␤1 and TGF-␤2, samples
were activated by adding 0.1 mL of 1 mol/L HCl for 10
minutes and neutralized with 100 ␮L of 1.2 mol/L
NaOH/0.5 mol/L HEPES before assay to measure the
total amount of TGF-␤.
VEGF ELISA
For 2-dimensional cocultures, fibroblasts and TE1
cells at a 1:1 ratio were seeded onto plates and cultured
with or without SB-505124 compound overnight. Supernatants were removed, and cells were washed with
DMEM. Cells were cultured for 48 hours with fresh culture
medium DMEM containing 2% FBS with or without the
compound. These supernatants were measured as samples using VEGF ELISA. For 3D culture, each sample was
cultured with or without SB-505124 compound for 48
hours, and these supernatants were measured using the
VEGF ELISA kit (R&D Systems).
Cell Proliferation Analysis
Cells were plated into a 96-well plate at a density
of 2.5 ⫻ 104 cells/mL and left to grow overnight. Cells
were treated with increasing concentrations of TGF-␤,
SB-505124, or GW788388 in triplicate. Proliferation was
analyzed using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay as previously
described.14
Statistics
Unless otherwise stated, all experiments show the
mean ⫾ SD of at least 3 independent experiments. Statistical significance was measured using the Student t
test, where P ⬍ .05 was judged to be significant.
Results
The Presence of Fibroblasts Is Required for
ESCC-Induced Angiogenesis
Studying the role of fibroblasts in ESCC-induced
network formation in vivo is technically challenging. To
overcome some of these issues, we developed a novel 3D
model of ESCC-induced angiogenesis, allowing us to
study the role of CAFs in vascular network formation13
(see Supplementary Figure 1 for scheme; see supplemental material online at www.gastrojournal.org). In the
model, factors derived from either fibroblasts or a mix-
FIBROBLASTS DRIVE ESCC–INDUCED ANGIOGENESIS
1985
ture of fibroblasts and ESCC cells cause HMVECs to
detach from the tissue culture plate and migrate upward
into the 3D collagen gel, where they organize and form
vascular networks. In an initial series of studies, it was
found that coculture of the ESCC lines with the HMVECs
(TE1, TE8, and TE11) did not lead to vascular network
formation (Figure 1A). However, coculture of the esophageal fibroblasts (FEF3) with the HMVECs led to endothelial cell migration and formation of moderately welldifferentiated vascular networks as shown by CD31 and
von Willebrand factor staining (Figure 1B and not
shown). An increase in the fibroblast concentration from
1 ⫻ 105 cells/mL to 1.5 ⫻ 105 and 2.5 ⫻ 105 cells/mL was
associated with increased vascular network organization
(Figure 1B). Addition of ESCC cells to the fibroblast/
HMVEC cocultures had the most striking effects on
vascular network formation and led to the establishment of very organized capillary-like structures (Figure
1C). Increasing the numbers of both the fibroblasts
and ESCC cells was associated with significantly more
organized capillary networks (Figure 1B, D, and E),
showing that the interaction of both the ESCC cells
and fibroblasts was critical for efficient vascular network formation.
Coculture of Esophageal Fibroblasts With
ESCC Cells Leads to Their Activation and
Transdifferentiation Into Myofibroblasts
CAFs are typically in an activated state, having
undergone transdifferentiation to a myofibroblast phenotype. Growing the human esophageal fibroblasts with
the ESCC line TE1 for 48 hours led to a change in
phenotype associated with the increased cytoplasmic expression of the myofibroblast marker ␣-SMA and fibroblast activation protein (Figure 2A and B and not shown).
Three additional esophageal carcinoma lines were also
noted to induce a similar degree of myofibroblast transdifferentiation (TE11, TE8, and TE12; Figure 2C and D).
The induction of the myofibroblast phenotype was induced through soluble factors derived from the ESCC
cells, as shown by the ability of conditioned media from
a panel of ESCC lines to induce ␣-SMA expression in the
fibroblasts (Figure 2E). In contrast, conditioned media
from primary human esophageal keratinocytes (EPC2)
were unable to induce ␣-SMA expression in the fibroblasts (Figure 2F).
4™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™
Figure 2. Coculture of esophageal fibroblasts with ESCC cells leads to their activation and transdifferentiation into myofibroblasts. (A) Control
GFP-tagged fibroblasts (green) expressed very little ␣-SMA. (B–D) Fibroblasts cocultured with the ESCC lines (TE1, TE11, TE8, TE10, and TE12) for
48 hours stain strongly for stress fibers of ␣-SMA (red). (E) Western blot analysis showing that incubation of fibroblasts with conditioned media (CM)
from ESCC lines for 47 hours leads to increased ␣-SMA expression in esophageal fibroblasts. Blots were stripped and reprobed with anti–␤-actin
as a loading control. (F) Conditioned media from human esophageal keratinocytes does not induce ␣-SMA expression in esophageal fibroblasts.
Scale bar ⫽ 100 ␮m.
BASIC–
ALIMENTARY TRACT
June 2008
1986
NOMA ET AL
Paracrine TGF-␤ Secreted From ESCC Cells
Is Responsible for Esophageal Fibroblast
Transactivation
ELISA experiments showed that all 4 ESCC lines
secreted high levels of both TGF-␤1 and TGF-␤2 (Figure
3A). To rule out the role of other possible growth factors,
further studies were performed showing that the ESCC
GASTROENTEROLOGY Vol. 134, No. 7
cells secreted very little platelet-derived growth factor or
insulin-like growth factor (data not shown). In a similar
manner to that of ESCC-conditioned media, exogenous
TGF-␤ was also shown to induce esophageal fibroblast
transdifferentiation associated with increased ␣-SMA expression (Figure 3B). Stimulation of the fibroblasts with
exogenous TGF-␤ was also accompanied by increased
BASIC–
ALIMENTARY TRACT
SMAD signaling, as shown by the rapid increase in phospho-SMAD2 (Figure 3B). Evidence for TGF-␤1 being the
ESCC-derived fibroblast-activating factor came from the
similar induction of nuclear SMAD3 accumulation seen
in both exogenous TGF-␤1 and ESCC conditioned media
(Figure 3C). Next, we investigated whether exogenous
TGF-␤ would stimulate the fibroblasts to drive vascular
network formation. Treatment of esophageal fibroblast/
HMVEC cocultures with 1 ng/mL TGF-␤ for 7 days led to
a statistically significant increase in capillary network
formation (Figure 3D) and showed the essential role of
TGF-␤–induced fibroblast differentiation in the angiogenic process. To show the essential role of the fibroblasts in TGF-␤–induced vascular network formation, we
treated 3D monocultures of endothelial cells with TGF-␤
(1 ng/mL) and observed very little vascular network formation. Indeed, the endothelial cells remained attached
to the plates and did not migrate upward into the collagen (Supplementary Figure 2; see supplemental material
online at www.gastrojournal.org).
TGF-␤ plays a complex role in tumor progression and
is known to be growth inhibitory to most epithelial cell
types. To overcome the effects of TGF-␤, most carcinoma
cells escape by down-regulating their expression of
TGF-␤ receptors and instead secrete autocrine/paracrine
TGF-␤ that recruits the surrounding stromal cells. In
agreement with this idea, the ESCC lines tested were not
found to express any TGF-␤ receptor subtype II protein,
whereas esophageal fibroblasts and the parent esophageal
keratinocyte line EPC2 maintained receptor expression
(Figure 3E). The ESCC lines were shown to have escaped the inhibitory effects of TGF-␤ and proliferated
normally in the presence of increasing concentrations
of TGF-␤ (0.1–10 ng/mL), whereas the primary human
esophageal keratinocyte line EPC2 was growth arrested
(Figure 3F).
The TGF-␤–Specific Inhibitor SB-505124
Inhibits ESCC-Induced Fibroblast
Transdifferentiation
Pharmacologic approaches that block tumor neoangiogenesis are an attractive therapeutic option. We
next tested whether the specific inhibitor of TGF-␤ re-
FIBROBLASTS DRIVE ESCC–INDUCED ANGIOGENESIS
1987
ceptor kinase SB-505124 blocked fibroblast transdifferentiation. Pretreating the esophageal fibroblasts with increasing concentrations of SB-505124 (0.01–10 ␮mol/L)
successfully blocked the TGF-␤–induced increases in
␣-SMA expression in the esophageal fibroblasts (Figure
4A). Maximal inhibition of the TGF-␤–induced fibroblast
activation was seen at 1 ␮mol/L SB-505124, and this concentration was selected for all subsequent experiments.
Treatment of the cells with SB-505124 also blocked ␣-SMA
induction and SMAD2 activation following treatment
with ESCC conditioned media (Figure 4B and C). To
investigate whether TGF-␤ inhibition was growth inhibitory on any of the cell types used in the neoangiogenesis
assay, endothelial cells (HMVEC), fibroblasts (FEF3), and
3 ESCC lines (TE1, TE8, and TE12) were treated with
increasing concentrations of each of 2 structurally distinct small molecule inhibitors of TGF-␤ (SB-505124 and
GW788388) for 72 hours. Measuring levels of proliferation using the MTT assay showed that neither TGF-␤
inhibitor affected the growth of any of the cell types
tested (Figure 4D and E).
The TGF-␤ Inhibitors SB-505124 and
GW788388 Inhibit ESCC-Induced Vascular
Network Formation
We next investigated whether inhibition of fibroblast activation blocked ESCC-induced vascular
network formation. Coculture of fibroblasts with the
HMVECs led to endothelial cell migration but very
little differentiated vascular network formation (Figure
5A). Treatment of the fibroblast/HMVEC cocultures with
the TGF-␤ inhibitor GW788388 (1 ␮mol/L) did not significantly alter the level of vascular network organization
(Figure 5A), showing that there was only limited fibroblast activation in the absence of ESCC cells. As before,
addition of the ESCC lines TE1 and TE8 to the fibroblast/HMVEC coculture dramatically increased the level
of vascular network organization (Figure 5B and C). In
contrast, treatment of the ESCC/fibroblast/HMVEC cocultures with either SB-505124 (1 ␮mol/L) or GW788388
(1 ␮mol/L) led to a complete reversal of vascular network
formation (Figure 5B and C). These results show that
inhibition of fibroblast activation by small molecule
4™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™™
Figure 3. ESCC lines secrete paracrine TGF-␤ that induces the transdifferentiation of esophageal fibroblasts. (A) ELISA assay showing secretion of
total TGF-␤1 and TGF-␤2 by esophageal cancer cell lines (TE). TE cells were cultured for 48 hours, and secreted TGF was measured by ELISA.
(B) Exogenous TGF-␤1 induces ␣-SMA expression in fibroblasts and stimulates TGF-related signaling pathways. Exogenous rhTGF-␤1 (48 hours)
induces ␣-SMA expression in esophageal fibroblasts. Exogenous rhTGF-␤1 induces SMAD2 phosphorylation in human esophageal fibroblasts.
(C) Both exogenous rhTGF-␤1 and ESCC conditioned media induce SMAD3 activation in esophageal fibroblasts. Fibroblasts were treated with either
rhTGF-␤1 (1 ng/mL) or conditioned medium of TE1 for 1 hour. Images show p-SMAD3 up-regulation and its subsequent nuclear translocation
(green), cell morphology is indicated by phalloidin (red), and nuclei are indicated by DAPI (blue). (D) Exogenous TGF-␤1 markedly enhances vascular
network formation in the absence of ESCC cells. Fibroblasts and HMVECs were cocultured in the presence of TGF-␤1 (1 ng/mL) for 7 days. Vascular
network formation was stained for CD31 (red), fibroblasts (GFP; green), and nuclei (DAPI; blue). (Inset) CD31 staining alone. The graph shows the
significantly (P ⬍ .005) increased numbers of microcapillary networks per field treated by TGF-␤1. (E) Western blot showing expression of TGF␤RII
expression in a panel of ESCC lines, fibroblasts, and primary human esophageal keratinocytes (EPC2). (F) Exogenous TGF-␤1 (72 hours) reduces the
growth of human esophageal keratinocytes (EPC2) but not ESCC lines. Cell proliferation was measured by the MTT assay. Images: scale bar ⫽ 50
␮m. (D) Scale bar ⫽ 200 ␮m.
BASIC–
ALIMENTARY TRACT
June 2008
1988
NOMA ET AL
GASTROENTEROLOGY Vol. 134, No. 7
Figure 4. Pharmacologic inhibitors of TGF-␤ signaling block ESCC-induced fibroblast transdifferentiation. (A) The TGF-␤ receptor kinase inhibitor
SB-505124 blocks TGF-␤–induced fibroblast transdifferentiation. Fibroblasts were cultured in the presence of TGF-␤1 (1 ng/mL) and increasing
concentrations of SB-505124 (0.01–10 ␮mol/L) for 48 hours, followed by blotting for ␣-SMA expression. (B) SB-505124 inhibits ␣-SMA over
expression in fibroblasts stimulated by ESCC-conditioned medium (CM; from TE1 cells). Fibroblasts were cultured with CM as described in Materials
and Methods in the absence or presence of SB-505124 (1 ␮mol/L) for 48 hours. (C) SB-505124 inhibits phosphorylation of SMAD2 induced by
ESCC-conditioned media. Fibroblasts were cultured with TGF-␤1 (1 ng/mL) or conditioned medium as indicated and treated with SB-505124 (1
␮mol/L) for 1 hour. (D and E) TGF-␤ inhibitors have little effect on the proliferation of ESCC cells, fibroblasts, or HMVECs. Fibroblasts (FEF3),
HMVECs, and esophageal cancer cell lines (TE) were treated with increasing concentrations of TGF␤RI inhibitors (either SB-505124 or GW788388)
for 72 hours before being subjected to the MTT assay. The results were evaluated as a percentage of control absorbance.
BASIC–
ALIMENTARY TRACT
TGF-␤ inhibitors blocks ESCC-induced neoangiogenesis (Figure 5D).
Pharmacologic Inhibition of TGF-␤ Inhibits
VEGF Secretion From Both Esophageal
Fibroblasts and ESCC Cells
VEGF is one of the most potent inducers of angiogenesis. To investigate the possible role of paracrine
VEGF release from esophageal fibroblasts in ESCC-induced neoangiogenesis, we tested VEGF release from
cocultured and TGF-␤–stimulated esophageal fibroblasts
using an ELISA. Unstimulated esophageal fibroblasts
produce a basal level of VEGF under 2-dimensional cell
culture conditions, and this was significantly increased
when the fibroblasts were grown under 3D conditions
(Figure 6A and B). Treatment of the fibroblasts with the
TGF-␤ inhibitor SB-505124 did not reduce basal levels of
VEGF under 2-dimensional cell culture conditions (data
not shown); however, it did inhibit basal VEGF release
when the fibroblasts were grown under 3D cell culture
conditions (Figure 6B). This showed that there was some
level of fibroblast activation when the cells were switched
from the 2-dimensional to 3D cell culture conditions.
Stimulation of the fibroblasts with exogenous TGF-␤ (1
ng/mL) led to a significant increase in the level of VEGF
secreted (Figure 6A). In a similar manner, coculture of the
fibroblasts with the ESCC line TE1 also led to a significant increase in VEGF secretion under both 2-dimensional and 3D cell culture conditions (Figure 6A and B).
Treatment of the ESCC/fibroblast cocultures with SB505124 (1 ␮mol/L) led to a significant inhibition in the
level of VEGF, showing a possible mechanism by which
ESCC-activated fibroblasts can induce vascular network
formation.
As a final test, we determined whether inhibition of
VEGFR2 signaling via the selective inhibitor GW654652
could also block ESCC-induced vascular network formation. Coculture of the HMVECs and fibroblasts led to
some limited vascular network formation after 7 days of
culture, and this was not affected by the presence of
GW654652 (1 ␮mol/L) (Figure 6C). Treatment of the
ESCC, fibroblast, and HMVEC cultures with GW654652
(1 ␮mol/L) did not completely inhibit vascular network
formation but did lead to a reduction in the number of
endothelial cell tubes formed (Figure 6C). Analysis of
these results showed a significant reduction (P ⬍ .005) in
the extent of ESCC-induced vascular network formation
following GW654652 treatment (Figure 6D). Similar results were also seen upon ESCC-induced vascular network formation following treatment with an antibody
directed against VEGF receptor (bevacizumab) (Supplementary Figure 3; see supplemental material online at
www.gastrojournal.org). A sample scheme showing how
ESCC cells stimulate fibroblasts to drive vascular network formation is shown in Figure 7.
Discussion
Here we show for the first time, using a novel 3D
in vitro model of the tumor microenvironment, the critical role of the host fibroblasts in directing ESCC-induced vascular network formation. This model differs
FIBROBLASTS DRIVE ESCC–INDUCED ANGIOGENESIS
1989
BASIC–
ALIMENTARY TRACT
June 2008
Figure 5. Pharmacologic inhibitors of TGF-␤ signaling block ESCC-induced vascular network formation. (A) TGF-␤ inhibition has little effect on fibroblast-induced
vascular network formation. Fibroblasts (FEF3; 1.5 ⫻ 105 cells/mL) were incubated with HMVECs in the absence or presence of 1 of 2 TGF-␤ inhibitors (SB-505124
or GW788388, both 1 ␮mol/L) for 7 days. Cultures were stained for HMVECs (CD31; red), fibroblasts (GFP; green), and nuclei (DAPI; blue). (B and C) TGF-␤
inhibition completely blocks ESCC-induced vascular network formation. Addition of either SB-505124 or GW788388 (both 1 ␮mol/L) led to complete inhibition of
vascular network formation. (D) Bar graph shows mean data for vascular network formation in the absence and presence of either SB-505124 or GW788388. All
representative images are shown as 3-color merges, and original monochrome CD31 (white) images are inset. Scale bar ⫽ 200 ␮m.
1990
NOMA ET AL
GASTROENTEROLOGY Vol. 134, No. 7
BASIC–
ALIMENTARY TRACT
Figure 6. Activated fibroblasts secrete VEGF, leading to increased vascular network formation. (A) Stimulation of esophageal fibroblasts with either
TGF-␤ through coculture with ESCC cells leads to enhanced VEGF release in 2-dimensional adherent culture. Fibroblasts (FEF3) in monoculture or
FEF3 and ESCC cells (TE1) at a ratio of 1:1 in coculture were cultured in the absence or presence of SB-505124 (1 ␮mol/L) for 48 hours.
Supernatants were harvested and quantified for VEGF expression using a specific ELISA. (B) Stimulation of esophageal fibroblasts with either TGF-␤
or ESCC conditioned media leads to enhanced VEGF release in 3D culture. Monocultures and cocultures of esophageal fibroblasts were grown in
a 3D collagen in the absence or presence of SB-505124 for 48 hours. (C) The VEGF inhibitor GW654652 (1 ␮mol/L) inhibits vascular network
formation. (D) The bar graph shows mean data for vascular network formation in the absence and presence of GW654652. All representative images
are shown as 3-color merges; monochrome images of CD31 are inset. Scale bar ⫽ 200 ␮m.
June 2008
FIBROBLASTS DRIVE ESCC–INDUCED ANGIOGENESIS
1991
from that in previous studies because it contains cancer
cells, fibroblasts, and endothelial cells. Coculture of
ESCC cells with HMVECs did not stimulate vascular
network formation in the absence of fibroblasts. Whereas
the coculture of fibroblasts with HMVECs induced some
endothelial cell migration into the collagen, it was associated with large, poorly defined vascular networks with
little organized branching. Truly organized vascular network formation was only seen when ESCC cells were
added to the fibroblast/HMVEC cultures. This suggested
both that the fibroblasts were absolutely required for
endothelial tube formation and that a prior fibroblast
activation step, via factor(s) released from the ESCC cells,
was required for efficient network formation. Activated
fibroblasts are often observed in the stroma associated
with growing tumors.15,16 In agreement with this observation, we noted that coculture of ESCC cells with esophageal fibroblasts led to their transdifferentiation into the
myofibroblast phenotype. Myofibroblasts are known to
be a good model for CAFs because the activated fibroblasts within the tumor milieu exhibit a phenotype that
is virtually indistinguishable from that of myofibroblasts.17
One of the best known factors responsible for the phenotypic switch of fibroblast to myofibroblast is TGF-␤.18
Loss of TGF-␤ receptor function is a common event in
cancer,18 and ESCC cells are no exception. They typically
become insensitive to the growth inhibitory effects of
TGF-␤ through either the loss of TGF-␤ receptor expression or the acquisition of missense mutations in
TGF␤RII.19 –21 In agreement with these published studies,
we found that our panel of ESCC lines was refractory to
the growth inhibitory effects of exogenous TGF-␤,
whereas primary human esophageal keratinocytes were
strongly growth inhibited. Loss of TGF␤RII expression in
the ESCC lines was also accompanied by the autocrine/
paracrine production of both TGF-␤1 and TGF-␤2.
Both ESCC conditioned media and exogenous TGF-␤
were similarly able to induce the myofibroblast phenotype and stimulate SMAD signaling, suggesting that
TGF-␤ was the factor likely for the fibroblast activation
observed. The requirement for TGF-␤–induced fibroblast
activation in neoangiogenesis was shown by the ability of
exogenous TGF-␤ to enhance vascular network formation in the absence of the ESCC cells. Further evidence
for the role of TGF-␤ in ESCC-induced fibroblast activation came from studies showing that a pharmacologic
inhibitor of TGF-␤ blocked the ability of ESCC-conditioned media induction of the myofibroblast phenotype
in esophageal fibroblasts.
Having shown that ESCC-secreted TGF-␤ is essential
for fibroblast-induced activation, we next tested whether
pharmacologic inhibition of TGF-␤ signaling could inhibit vascular network formation. For these studies we
used SB-505124, a selective TGF-␤ receptor antagonist
with very little activity against any other kinase tested.22
Increasing concentrations of SB-505124 were found to
inhibit myofibroblast transdifferentiation induced both
by TGF-␤ and ESCC conditioned media. The essential
role of TGF-␤–induced fibroblast activation in vascular
BASIC–
ALIMENTARY TRACT
Figure 7. Schematic illustration showing the role of ESCC cells and fibroblasts in vascular network formation.
Esophageal cancer cells produce TGF-␤
to activate stromal normal fibroblasts.
Tumor stromal fibroblasts become
transdifferentiated into myofibroblasts
that secrete VEGF, which in turn induces endothelial cell migration and the
formation of a microcapillary network.
1992
NOMA ET AL
BASIC–
ALIMENTARY TRACT
network formation was indicated by the fact that SB505124 and the structurally unrelated TGF-␤ inhibitor
GW788388 significantly inhibited ESCC-induced neoangiogenesis. The requirement for both ESCC cells and
fibroblasts in the angiogenic process was shown by the
fact that neither of the TGF-␤ inhibitors tested blocked
the limited amount of vascular network formation induced by fibroblasts alone.
ESCC-induced fibroblast activation was shown to drive
vascular network formation through the stimulation of
VEGF release. VEGF is a potent proangiogenic factor that
stimulates endothelial cell migration and proliferation
and regulates microvascular permeability.23 There is compelling evidence that fibroblasts can be a major source of
growth factors that may contribute to tumor progression
and possibly angiogenesis.9,17,24 Studies using transgenic
mice expressing GFP under the control of VEGF promoter showed strong GFP staining in the stroma of
spontaneously arising mammary tumors.10 Here we show
that fibroblasts activated by exogenous TGF-␤ through
coculture with ESCC cells secrete significantly higher
VEGF levels through a mechanism involving SMAD signaling. The fact that a selective inhibitor of VEGF receptor signaling blocks efficient ESCC-induced vascular network formation provides the link between ESCC-induced
fibroblast activation and angiogenesis in this model. In
agreement with this, others have also shown that fibroblasts can release VEGF in response to hypoxic conditions25,26 and exogenous TGF-␤.27,28 It is unclear whether
the ESCC cells are releasing latent VEGF from the fibroblasts, as has been suggested by others,29 or are instead
driving de novo protein expression.
Here, we have shown that the stromal fibroblasts are
essential for the angiogenic response in ESCC, opening
up an intriguing new possibility for therapy. There is a
growing body of evidence suggesting that therapies targeted against angiogenesis can lead to dramatic responses in
diseases such as colorectal carcinoma, particularly when
combined with established chemotherapeutic regimens.30
Because ESCC cells are typically detected at relatively
advanced stages, optimized antiangiogenic/chemotherapy combinations are ideal novel therapeutic candidates.
The postulated role of CAFs in tumor invasion also makes
targeting the stromal fibroblast activation through the inhibition of TGF-␤ signaling an appealing therapeutic option.
Supplementary Data
Note: To access the supplementary material
accompanying this article, visit the online version of
Gastroenterology at www.gastrojournal.org, and at doi:
10.1053/j.gastro.2008.02.061.
References
1. Bissell MJ, Radisky D. Putting tumours in context. Nat Rev Cancer
2001;1:46 –54.
GASTROENTEROLOGY Vol. 134, No. 7
2. Dvorak HF. Tumors: wounds that do not heal. Similarities between tumor stroma generation and wound healing. N Engl J Med
1986;315:1650 –1659.
3. Elenbaas B, Weinberg RA. Heterotypic signaling between epithelial tumor cells and fibroblasts in carcinoma formation. Exp Cell
Res 2001;264:169 –184.
4. Nakayama H, Enzan H, Miyazaki E, et al. The role of myofibroblasts at the tumor border of invasive colorectal adenocarcinomas. Jpn J Clin Oncol 1998;28:615– 620.
5. Tsukamoto H, Mishima Y, Hayashibe K, et al. Alpha-smooth
muscle actin expression in tumor and stromal cells of benign and
malignant human pigment cell tumors. J Invest Dermatol 1992;
98:116 –120.
6. De Wever O, Mareel M. Role of tissue stroma in cancer cell
invasion. J Pathol 2003;200:429 – 447.
7. Direkze NC, Hodivala-Dilke K, Jeffery R, et al. Bone marrow contribution to tumor-associated myofibroblasts and fibroblasts. Cancer Res 2004;64:8492– 8495.
8. Sappino AP, Skalli O, Jackson B, et al. Smooth-muscle differentiation in stromal cells of malignant and non-malignant breast
tissues. Int J Cancer 1988;41:707–712.
9. De Wever O, Nguyen QD, Van Hoorde L, et al. Tenascin-C and
SF/HGF produced by myofibroblasts in vitro provide convergent
proinvasive signals to human colon cancer cells through RhoA
and Rac. FASEB J 2004;18:1016 –1018.
10. Fukumura D, Xavier R, Sugiura T, et al. Tumor induction of VEGF
promoter activity in stromal cells. Cell 1998;94:715–725.
11. Lioni M, Brafford P, Andl C, et al. Dysregulation of claudin-7 leads
to loss of E-cadherin expression and the increased invasion of
esophageal squamous cell carcinoma cells. Am J Pathol 2007;
170:709 –721.
12. Andl CD, Mizushima T, Nakagawa H, et al. Epidermal growth
factor receptor mediates increased cell proliferation, migration,
and aggregation in esophageal keratinocytes in vitro and in vivo.
J Biol Chem 2003;278:1824 –1830.
13. Velazquez OC, Snyder R, Liu ZJ, et al. Fibroblast-dependent differentiation of human microvascular endothelial cells into capillary-like, three-dimensional networks. FASEB J 2002;16:1316 –
1318.
14. Smalley KS, Contractor R, Haass NK, et al. Ki67 expression
levels are a better marker of reduced melanoma growth following
MEK inhibitor treatment than phospho-ERK levels. Br J Cancer
2007;96:445– 449.
15. Orimo A, Weinberg RA. Stromal fibroblasts in cancer: a novel
tumor-promoting cell type. Cell Cycle 2006;5:1597–1601.
16. Kalluri R, Zeisberg M. Fibroblasts in cancer. Nat Rev Cancer
2006;6:392– 401.
17. Orimo A, Gupta PB, Sgroi DC, et al. Stromal fibroblasts present in
invasive human breast carcinomas promote tumor growth and
angiogenesis through elevated SDF-1/CXCL12 secretion. Cell
2005;121:335–348.
18. Levy L, Hill CS. Alterations in components of the TGF-beta superfamily signaling pathways in human cancer. Cytokine Growth
Factor Rev 2006;17:41–58.
19. Tanaka S, Mori M, Mafune K, et al. A dominant negative mutation
of transforming growth factor-beta receptor type II gene in microsatellite stable oesophageal carcinoma. Br J Cancer 2000;82:
1557–1560.
20. Garrigue-Antar L, Souza RF, Vellucci VF, et al. Loss of transforming growth factor-beta type II receptor gene expression in primary
human esophageal cancer. Lab Invest 1996;75:263–272.
21. Fukai Y, Fukuchi M, Masuda N, et al. Reduced expression of
transforming growth factor-beta receptors is an unfavorable prognostic factor in human esophageal squamous cell carcinoma. Int
J Cancer 2003;104:161–166.
22. DaCosta Byfield S, Major C, Laping NJ, et al. SB-505124 is a
selective inhibitor of transforming growth factor-beta type I receptors ALK4, ALK5, and ALK7. Mol Pharmacol 2004;65:744 –752.
23. Dvorak HF, Brown LF, Detmar M, et al. Vascular permeability
factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis. Am J Pathol 1995;146:1029 –
1039.
24. Dong J, Grunstein J, Tejada M, et al. VEGF-null cells require
PDGFR alpha signaling-mediated stromal fibroblast recruitment
for tumorigenesis. EMBO J 2004;23:2800 –2810.
25. Hlatky L, Tsionou C, Hahnfeldt P, et al. Mammary fibroblasts may
influence breast tumor angiogenesis via hypoxia-induced vascular endothelial growth factor up-regulation and protein expression. Cancer Res 1994;54:6083– 6086.
26. Nakagawa T, Lan HY, Zhu HJ, et al. Differential regulation of VEGF
by TGF-beta and hypoxia in rat proximal tubular cells. Am J Physiol
Renal Physiol 2004;287:F658 –F664.
27. Kobayashi T, Liu X, Wen FQ, et al. Smad3 mediates TGF-beta1
induction of VEGF production in lung fibroblasts. Biochem Biophys Res Commun 2005;327:393–398.
FIBROBLASTS DRIVE ESCC–INDUCED ANGIOGENESIS
1993
28. Renner U, Lohrer P, Schaaf L, et al. Transforming growth factorbeta stimulates vascular endothelial growth factor production by
folliculostellate pituitary cells. Endocrinology 2002;143:3759 –
3765.
29. Ito TK, Ishii G, Chiba H, et al. The VEGF angiogenic switch of
fibroblasts is regulated by MMP-7 from cancer cells. Oncogene
2007;26:7194 –7203.
30. Jain RK. Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy. Science 2005;307:58 – 62.
Received January 17, 2008. Accepted February 19, 2008.
Address requests for reprints to: Meenhard Herlyn, DVM, or Keiran
S. M. Smalley, PhD, The Wistar Institute, 3601 Spruce Street, Philadelphia, Pennsylvania 19104. e-mail: [email protected] or ksmalley
@wistar.org
Supported by National Cancer Institute grant P01-CA098101 (to
M.H.).
The authors declare no financial conflicts of interest.
BASIC–
ALIMENTARY TRACT
June 2008